Silver Nanoparticles for Chronic Wound Management: Comparison
Please note this is a comparison between Version 3 by Camila Xu and Version 2 by Sabarees Govindaraj.
 
Abstract
Infections are the primary cause of death from burns and diabetic wounds. The clinical difficulty of treating wound infections with conventional antibiotics has progressively increased and reached a critical level, necessitating a paradigm change for enhanced chronic wound care. The most prevalent bacterium linked with these infections is 
Infections are the primary cause of death from burns and diabetic wounds. The clinical difficulty of treating wound infections with conventional antibiotics has progressively increased and reached a critical level, necessitating a paradigm change for enhanced chronic wound care. The most prevalent bacterium linked with these infections is
Staphylococcus aureus
, and the advent of community-associated methicillin-resistant 
Staphylococcus aureus has posed a substantial therapeutic challenge. Most existing wound dressings are ineffective and suffer from constraints such as insufficient antibacterial activity, toxicity, failure to supply enough moisture to the wound, and poor mechanical performance. Using ineffective wound dressings might prolong the healing process of a wound. To meet this requirement, nanoscale scaffolds with their desirable qualities, which include the potential to distribute bioactive agents, a large surface area, enhanced mechanical capabilities, the ability to imitate the extracellular matrix (ECM), and high porosity, have attracted considerable interest. The incorporation of nanoparticles into nanofiber scaffolds constitutes a novel approach to “nanoparticle dressing” that has acquired significant popularity for wound healing. Due to their remarkable antibacterial capabilities, silver nanoparticles are attractive materials for wound healing. This review focuses on the therapeutic applications of nanofiber wound dressings containing Ag-NPs and their potential to revolutionize wound healing.
has posed a substantial therapeutic challenge. Most existing wound dressings are ineffective and suffer from constraints such as insufficient antibacterial activity, toxicity, failure to supply enough moisture to the wound, and poor mechanical performance. Using ineffective wound dressings might prolong the healing process of a wound. To meet this requirement, nanoscale scaffolds with their desirable qualities, which include the potential to distribute bioactive agents, a large surface area, enhanced mechanical capabilities, the ability to imitate the extracellular matrix (ECM), and high porosity, have attracted considerable interest. The incorporation of nanoparticles into nanofiber scaffolds constitutes a novel approach to “nanoparticle dressing” that has acquired significant popularity for wound healing. Due to their remarkable antibacterial capabilities, silver nanoparticles are attractive materials for wound healing. 
  • composite nanofibers
  • silver nanoparticles
  • wound dressing

1. Brief Medical History of Silver Nanoparticles

Silver nanoparticles have been extensively used in various medicinal applications, as shown in (Figure 31). Silver nanoparticles are already widely used in wound dressings and burn treatment in biomedicine and also in the food and textile industries, in paints, household items, catheters, implants, and cosmetics, as well as in combination with a variety of materials to prevent infection [20,21,22,23,24,25][1][2][3][4][5][6]. During World War I, they treated soldiers’ wounds with silver leaves to stop infections and help them heal [26,27][7][8]. Silver as an ion was used in earlier civilizations, particularly in Egypt. This silver ion was used mainly in wound dressings to treat wounds that were hard to heal. In 1998, Ziehl-Abegg was the first firm to introduce AgNPs into wound dressings, resulting in the AgNPs antimicrobial dressing ActicoatTM. Silver products improve efficacy compared to standard wound dressing [28,29][9][10]. Acticoat and Actisorb, two silver-based wound dressings, are commercially available [30][11]. Dermatology increasingly employs metal nanoparticles to expedite wound healing and to treat and prevent bacterial infections [31][12].
Figure 31. Biomedical applications of silver nanoparticles.
Indeed, AgNPs’ excellent antimicrobial properties have already been tested against 650 bacterial strains [32,33,34][13][14][15]. Because of their extensive antimicrobial properties and ability to reduce the chance of infection from antibiotic-resistant strains, silver nanoparticles also found their application in wound management products [35,36,37,38,39][16][17][18][19][20]. AgNPs are utilized in wound care to prevent secondary infections since they are effective against a spectrum of microbes that can slow down the healing process [40][21]. Their size and charge allow them to enter microorganisms [41,42,43][22][23][24]. The AgNPs destroy many multi-resistant strains. It also helps rid pathogenic microbes that can slow or stop the typical stages of wound healing [44,45][25][26].
The biosynthesis of AgNP utilizing aqueous Bryonia laciniosa leaf extract resulted in rapid wound epithelialization and scarless wound repair without significant inflammation due to effective cytokine modulation. It was used as a wound healing agent due to its outstanding anti-inflammatory and antibacterial properties [46][27]. Biosynthesized AgNPs are made from Delonix elata leaf aqueous extract for wound healing in human patients who have had anorectal surgery [47][28]. Due to their sustained ability to release silver ions, which exhibit concentration-dependent toxicity in HaCaT cells, these nanoparticles have a high potential for usage in dermatology and wound healing [48][29]. Because AgNPs are biocompatible and can avoid this involuntary inflammatory action, they are not nullified by the immune system; thus, they could be used as anti-inflammatory agents [49][30]. Because of their anti-inflammatory properties, topical application of AgNPs at the wound site minimizes the release of inflammatory cytokines, lymphocytes, and mast cell infiltration, promoting wound healing with minimal scarring. A. Hebeish et al. also evaluated the anti-inflammatory effects of Ag by comparing them to indomethacin. This commercially available anti-inflammatory drug revealed a dose-dependent reduction in inflammation inside the rat bow edema model [50][31].
A study showed that AgNPs eradicate 44 strains of six fungi [51][32]. Gajbhiye et al. discovered that biogenic AgNPs were effective against Fusarium semitectumPleospora herbarumTrichoderma spp, Phoma glomerata, and Candida albicans. They also reported a synergic effect of AgNPs in conjunction with fluconazole [52,53][33][34]. When AgNPs were added, they changed the growth rates of all tested fungi except Mortierella spp, meaning that Chaetomium and Stachybotrys could not grow on gypsum products.
J.L. Speshock et al. investigated AgNPs’ potential in prokaryotic and eukaryotic organisms, and AgNPs at about 25 nm or even less were found to have exceptional potential for viral infection suppression [54][35]. AgNPs inhibited virus attachment, cell penetration, and the cell’s ability to propagate the virus [55][36]. As an HIV-1 antiviral nanohybrid and in the deactivation of SARS-Cov-2 spike proteins, TPU-Ag worked better than PVA-Ag. TPU-Ag and PVA-Ag nanofibrous membranes displayed increased antibacterial activity by increasing Ag content from 2 to 4 wt. Additionally, the developed membranes showed good mechanical and physical properties and antiviral and antibacterial activities [56][37].
Silver synthesized from M. Domestica extracts significantly affected breast cancer MCF-7 cells. In contrast, silver synthesized from O. Vulgare aqueous extracts had a dose-dependent effect on the A549 cell line [57,58][38][39]Moringa olifera stem bark extract was used to produce AgNPs. K. Vasanth et al. investigated the anticancer properties of these biosynthesized AgNPs. The flow cytometry analysis indicated that ROS generation caused apoptosis in HeLa cells [59][40]. According to the findings, AgNPs effectively prevent the development of HepG2 cells by inducing apoptosis [60][41]. Venkatesan et al. found that the human breast cancer cells MDA-MB-231 were killed by chitosan-alginate-biosynthesized AgNPs that were highly permeable (IC50 = 4.6 mg) [61][42]. A recent study found that packed quinazolinone polypyrrole with chitosan silver chloride nanocomposite was active against Ehrlich ascites carcinoma cells [62][43]. I.M. El-Sherbiny et al. found that Chitosan-silver hybrid nanoparticles cause HepG2 cells to die by turning down the BCL2 gene and the P53 gene [63][44]. A significant decrease in cyclobutene-pyrimidine-dimer creation demonstrated their chemo-preventive efficacy in HaCaT cells after UVB-induced DNA damage, which has a good potential for avoiding skin cancer [64][45]. The UVB-protective effectiveness of AgNPs in human keratinocytes is proportional to their size [65][46]. As a result, pre-treating HaCaT cells with smaller AgNPs (10–40 nm) helped shield skin cells from UVB-induced DNA damage and UV-induced apoptosis. Using 60 and 100 nm AgNPs, no prevention was obtained. AgNPs are increasingly used in healthcare and consumer products, so many commercial products now include these nanoparticles for topical administration to human skin.
Over one million people die from malaria yearly, caused by protozoal vector-borne diseases, the most prevalent and dangerous infections in wealthy nations [66][47]. Z. Jiang et al. are creating novel antimalarial strategies to control the malaria vector. AgNPs were tested against Plasmodium falciparum malarial parasites and other antimalarial medications [67][48]. The bio-reduction of AgNPs was 5%. The malaria vector Anopheles stephensi and chloroquine-sensitive and resistant P. falciparum strains were all treated with Cassia occidentalis leaf broth [68][49].

2. Wound Healing Properties of Silver Nanoparticles

Compounds of silver, such as silver nitrate and silver sulfadiazine, are often used to treat infections in chronic wounds and burns [69][50]. AgNPs help fibroblasts change into myofibroblasts, which makes wounds tighter and speeds up the healing of diabetic wounds. AgNPs accelerate wound healing by enhancing keratinocyte proliferation and migration [70,71][51][52]. AgNPs may engage with sulphur-containing proteins in bacterial membrane cells and, ideally, attack the respiratory chain, resulting in apoptosis [72][53]. When it comes into contact with the injured region, they cause neutrophil apoptosis by lowering mitochondrial function, which reduces cytokine production. As a result, the inflammatory response is modulated or reduced, resulting in faster healing [73,74][54][55]. However, because of their small size, AgNPs can easily penetrate biofilms and cell membranes, causing DNA damage, inhibiting cell proliferation, and inhibiting cellular ATP production [75][56]. The silver nanoparticles change the amount of m-RNA in the wound environment. Aside from antibacterial activities, silver surgical textiles exhibit an increase in healing properties; as an outcome, silver exploitation has an optimistic effect on cell migration and proliferation quality [76,77,78][57][58][59]. Cytokine modulation is mediated by silver nanoparticles’ anti-inflammatory activity [79][60]. As stated in the preceding section, Cytokines can stimulate fibroblasts and chondrocytes to generate ROS [80][61]. Thus, silver nanoparticle modulation of cytokine production can reduce ROS levels to avoid severe cellular damage and lag wound healing [81][62]. Silver has many antibacterial effects, making it less likely that bacteria will become resistant and more effective against microorganisms resistant to multiple drugs. When the amount of AgNPs in the dressing increases, the wound area becomes smaller and more collagen is deposited, which is linked to macrophage and fibroblast migration [82,83][63][64]. Sustained release mechanisms can decrease silver ion toxicity and stimulate local antibacterial activity [84,85][65][66].

3. Mechanistic Understanding of Silver Nanoparticles (AgNPs)

The usual quantity of silver in human plasma is less than 2 µg/mL, and this concentration comes from diet and particulate matter inhalation. Oral exposure to silver can also come via dietary supplements, contaminated water, or from eating fish and other aquatic species [86][67]. Ionic silver can be ingested orally, inhaled, or absorbed through wounds to enter the body. AgNPs are believed to be transported inside the body by two processes: pinocytosis and endocytosis. The development of a revolutionary medication delivery method was prompted by the discovery that nanoscale particles penetrate far deeper than bulk particles. Although the precise mode of action of AgNPs is not yet known, numerous ideas for their antibacterial qualities have been put forth. Its antibacterial effect is thought to solely be caused by the ongoing release of silver in its ionic state [87,88,89][68][69][70]. Silver ions cling to the cytoplasmic membrane and cell wall because of the sulphur protein affinity and electrostatic attraction. This increases the permeability of the membrane and causes the bacterial cell to rupture and degenerate. Reactive oxygen species are produced, and the respiratory enzymes are essentially deactivated when the silver ion enters the bacterial cell. Reactive oxygen species, a critical element in the mechanism of action for silver, contribute significantly to the disruption of the cell membrane and DNA damage (by interacting with sulphur and phosphorus in the DNA molecule), which hinder replication and reproduction and ultimately lead to microbe death. By denaturing ribosomes, silver ions also prevent the synthesis of ATP and hinder the formation of proteins. After anchoring and observing the cell’s surface, silver nanoparticles build up in the cellular wall pits of microorganisms, causing the denaturation and degeneration of the cell membrane. Due to their micro size, they can easily enter cells, rupturing cell organelles and even causing cell lysis. They interfere with the phosphorylation of protein substrates, which can cause cell death and proliferation, which has an impact on the bacterial transduction process as well. Due to their cellular walls being shorter than those of Gram-positive bacteria, Gram-negative bacterial strains are more susceptible to the effects of AgNPs [86,89][67][70]. Silver nanoparticles have a significant downside in that bacterial biofilms make them less effective and penetrating. Due to their intricate structure, biofilms typically change the transport chain to shield the membrane from both silver ions and nanoparticles. The nanoparticle size, which is around 50 nm, severely obstructs the path of penetration that is currently being used. Additionally, it has been observed that silver nanoparticle adsorption and deposition on bacterial biofilms reduces the nanoparticles’ ability to diffuse into bacterial cells (Figure 42). Silver’s interaction with a molecule containing a thiol group in bacterial, fungal, and fungus cells provides the basis for silver nanoparticles’ antibacterial effect (Figure 42). It has been observed that bacterial and fungal cells undergo structural changes after coming into touch with silver nanoparticles, albeit the precise process is yet unclear. Silver nanoparticles have higher antibacterial and antifungal characteristics than normal silver particles because of their extensive surface area, which enables better interaction with bacterial and fungal pathogens. Additionally, the gel made of silver nanoparticles penetrates bacteria and fungi in addition to attaching to cell membranes. Silver penetrates cells and connects to the cell membrane and wall, which prevents the cell from respiring [90,91][71][72]. When silver is present, Escherichia coli is prevented from absorbing phosphate and from releasing mannitol, succinate, proline, and glutamine. As a result, silver nanoparticles can be used as effective growth inhibitors in a wide range of microbes and are helpful in many antibacterial control systems [92,93][73][74].
Figure 42. Antibacterial mechanism of silver nanoparticles.

4. Silver Nanoparticles and Their Synthesis

 
Figure 5. Different approaches of silver nanoparticles synthesis.
New alternative techniques that are cost-efficient, energy-efficient, and environmentally benign are swiftly becoming available to counteract these negative environmental consequences. There has been a thorough literature assessment on the development of green or biological synthesis [98,99,100,101][75][76][77][78]. In green synthesis, an ecologically friendly substance such as microbial (fungal and bacterial) enzymes and phytochemicals from plant extracts are used instead of the capping/stabilizing and reducing agents used in chemical reduction procedures (leaves, roots, barks, flowers, fruits, peels, and seeds). These biological processes generate biocompatible nanoparticles suitable for pharmaceutical and biomedical applications [102,103][79][80]. While utilizing microbes as reducing agents in nanoparticle manufacturing is quite tricky and involves several methods, it is more advantageous than chemical and physical techniques. Because they are widely accessible, simple to extract, and do not need laborious processes, plant extracts are considered a solution to the issues mentioned above in the manufacturing of AgNP. For the most part, because they are so readily available, plant extracts are employed to make nanoparticles. Manufacturing AgNPs with regular shapes and sizes is the main difficulty with plant-based synthesis, and the reduction processes of biological approaches are not well understood [104,105,106,107,108][81][82][83][84][85].

5. Electrospinning

An electrospinning machine consists of four main parts: a high-power source, a syringe pump, a syringe needle carrying solutions, and a fiber deposition collector, as shown in (Figure 63). A positive electrode is attached to the needle, and a negative electrode is connected to the collector to create the applied electric field. As a result, the repulsion charge accumulates near the hemispherical needle tip when a voltage is applied. A Taylor cone is made when the repulsive charge surpasses the surface tension. The negative electrode, which serves as the collector in this procedure, is where the polymer solution is directed to create fibers. The polymer solution evaporates, leaving behind dry fibers ranging in size from nanometers to micrometers deposited on the collector [109,110][86][87].
Figure 3. A schematic diagram of electrospinning apparatus.

6. Advantages of Silver and Fibre Platforms

The ideal wound dressing should fulfill a number of criteria, including those listed below: (i) representing a physical barrier that is permeable to oxygen while also maintaining or providing a moist environment; (ii) being sterile, non-toxic, and protective against microorganism infections; (iii) providing an appropriate tissue temperature to favour epidermal migration and promote angiogenesis; and (iv) being non-adherent to prevent traumatic removal after healing. An ideal wound dressing should have all the qualities listed above, but it is challenging for one kind of dressing to meet every one of these needs. Creating a moist wound environment reduces dehydration and cell death. It facilitates angiogenesis and epidermal migration. It preserves moisture at the site of the wound. Excess exudate must be removed for the wound to heal, but it can also cause healthy tissue to macerate, creating a persistent wound. It enables gaseous exchange. Oxygenation regulates exudate levels and promotes fibroblast and epithelial growth. It prevents infection by prolonging the inflammatory phase and preventing epidermal migration and collagen formation; microbial infections slow the healing of wounds. Low adherence and painless removal of adherent dressings can be uncomfortable and can worsen existing granulation tissue damage. The cost-effective and optimal dressing should promote wound healing while remaining reasonably priced [128,129,130][88][89][90]. The main categories of wound-dressing materials are fibers, gels, membranes, films, sponges, and hydrocolloids, as shown in (Table 21). Nanofiber mats are a superior option for drug delivery compared to all other biomaterials because of their numerous advantages and inherent properties, as shown in (Table 21). The incorporation of nanoparticles into nanofiber scaffolds constitutes a novel approach to “nanoparticle dressing” that has acquired significant popularity for wound healing (Table 21). Due to their remarkable antibacterial capabilities, silver nanoparticles are attractive materials for wound healing. Numerous wound-dressing materials have been created in this area (Table 32), based either on synthetic or natural polymers.
Table 1. The benefits and drawbacks of the various types of nanomaterials.
S. No.Wound Dressing MaterialsSize of AgNPs (nm)Target MicrobeIn Vivo/In Vitro ModelAdvantage of NanocoatingRef.
1Fibers
  • nonadherent, nontoxic, nonallergenic
  • allow gaseous exchange
  • remove excess exudates
  • barrier against microbes
  • sustain release
  • maintain humidity
  • tensile strength
  • increase bioavailability
  • fibroblast attachment and proliferation
  • keratinocyte attachment and proliferation
  • tunable porosity
  • ECM mimicking
  • bio-compatibility
  • electro-catalytic properties
  • thermal conductivity
  • electrical conductivity
  • structural stability
  • loading efficiency
  • high surface area to volume ratio
  • mechanical strength
  • unsuitable for third degree, eschar, and dry wounds
  • if the wound is highly exudative, need a secondary dressing
[91]
2Membranes
  • act as physical barriers
  • membranes simulate extracellular matrix (ECM) structure
  • assure gas exchange, cell proliferation, and nutrient supply
  • the materials and solvents used in the production process may be harmful
[92]
3Films
  • impermeable to bacteria
  • allows the healing process to be monitored
  • painless removal
  • hard to handle
  • non-absorbent
  • adhere to the wound bed and cause exudate accumulation
[93]
4Hydrocolloids
  • non-adherent
  • high density
  • painless removal
  • high absorption properties
  • can be cytotoxic
  • have an unpleasant odor
3Chitosan/Poly (Ethylene Oxide) matrix5E. coli-AgNPs, because of their size and structure, were found to increase antibacterial activity when introduced.[99]
  • low mechanical stability
  • maintain acidic pH at the wound site
4Chitin/nanosilver composite scaffolds5 nmE. coli and [94]
S. aureusL929The scaffolds are antibacterial and have excellent blood clotting capabilities, which will help with wound healing. These scaffolds were hazardous to mouse fibroblasts in vitro. Whether in vitro cytotoxicity affects in vivo wound healing is unknown.[100]5Hydrogels
  • high absorption properties
  • provide a moist environment at the wound site
  • water retention
  • oxygen permeability
  • ensure the solubility of growth factor/antimicrobial agents
  • weak mechanical properties
  • need a secondary dressing
5Activated Carbon coated silver nanocomposite50–400S. aureus, Klebsiella pneumoniae and P. aeruginosa[95]
-When compared to plain activated carbon, the Ag composites’ antibacterial activity was significantly higher.[101]6Sponges
  • high porosity
  • thermal insulation
  • sustain a moist environment
1Chitosan-Poly Vinyl Pyrrolidone (PVP) composite10–30E. coli and S. aureusL929 cell lineCompared to the control sample, silver nanocomposite reduced the amount of inflammatory cells by 99.[97]
  • absorb wound exudates
  • enhance tissue regeneration
  • mechanically weak
  • may provoke skin maceration
  • unsuitable for third degree burn treatment or wounds with dry eschar
[96]
Table 2. A summary of available AgNP-based wound dressing products and their benefits.
2Plumbagin caged AgNP-collagen scaffolds60 nmE. coli and B. subtiliswistar rat/Swiss 3T6The antibacterial and wound-healing capabilities of silver and plumbagin in the PCSN cross-linked collagen scaffold showed the importance of nano-biotechnology.[98]
6
Silver nano-coatings on cotton gauzes100–300 nmS. aureusHaCaT/3T3The developed textile materials show promise as an alternative to traditional wound dressings due to their antimicrobial properties and biocompatibility.[102]
7Polyurethane Foam mixed Ag-NPs Dressing100E. coliP. aeruginosa and S. aureusHuman fibroblastWound healing was enhanced by the use of the foam dressing.[103]
8AgNP gelatin hydrogel pads7.7–10.8 nmE. coliS. aureus P. aeruginosaHuman’s normal skin fibroblastsGelatin hydrogel pads infused with silver nanoparticles have shown promise as antibacterial wound dressings.[104]
9Chitosan-PEG hydrogel75E. coliP. aeruginosa and S. aureusRabbitOn day 14, the dermal layer of skin and the collagen pattern were both healthy in the Ag-NPs impregnated chitosan-PEG hydrogel group.[105]
10AgNPs incorporated Pluronic F127 and Pluronic F68 thermosensitive gel-E. coliS. aureus and P. aeruginosa-Gel may disrupt the structure of bacterial cell membranes, allowing the substance to enter the cell, where it can condense DNA, combine and coagulate with the cytoplasm, and ultimately kill the bacteria by causing the cytoplasmic component to leak out.[106]
11Chitosan nanofiber25S. aureusWistar Hannover ratsBiological media had a substantial impact on the release of silver; proteins blocked the release of the metal, whereas inorganic ions slowed it down. As a result, to elicit in vivo antibacterial activities, a high concentration of AgNPs was required.[107]
12Asymmetric Wettable Chitosan nanocomposite25E. coliP. aeruginosa and S. aureusHEK293 cell lineThe dressing has been shown to encourage cell growth in an in vitro cytocompatibility study.[108]
13Cellulose hydrogel5–50E. coli and S. aureusNew Zealand rabbitThree days faster wound healing was seen using nanohydrogel compared to the control group.[109]
14Chitosan gels15P. aeruginosaHuman dermal fibroblastsChitosan gels containing AgNPs showed improvement in biocompatibility tests on primary fibroblasts.[110]
15Silk fibroin/ carboxymethyl chitosan composite sponge4.9 ± 1.9 nmS. aureus and P. aeruginosa-This AgNP-loaded SF/CMC sponge shows promise as a potential antimicrobial wound dressing.[111]
16Chitosan cross-linked bilayer nanocomposite45E. coliP. aeruginosa and S. aureusL929 cell lineThe treated group’s organized and developed epithelium was a marked improvement over that of the control group.[112]
17AgNPs/Bacterial cellulose composites10–30 nmE. coliS. aureus and P. aeruginosaEpidermal cellsIn vitro studies show that a nanostructured AgNP-BC gel-membrane has the potential to be an effective antimicrobial wound dressing with good biocompatibility for the expedited healing of scald wounds.[113]
18Silver NPs embedded bacterial cellulose gel membranes30S. aureusWestar ratsA significant amount of healing (85.92%) occurred after 14 days of treatment.[114]
19β-chitin-based hydrogels5E. coli and S. aureusERO cell lineManufactured scaffolds showed improved whole-blood clotting ability.[115]
20Silver Alginate/Nicotinamide Nanocomposites20–80E. coli and S. aureusMiceSignificant wound healing had occurred by the fourth day of treatment.[116]
21Hyaluronan Nanofiber25E. coli and S. aureusCell line (NIH 3T3)Since nanoparticles are so much smaller than typical particles, they are able to exert a far stronger effect on microbes.[117]
22Chitosan-Ag/ZnO composite dressing10–30 nmDrug sensitive E. coliS. aureus and P. aeruginosaBALB/c mice /L02 cellsThese findings support the feasibility of using the prepared chitosan-Ag/ZnO composite dressing in wound care.[118]
23Chitosan-based multifunctional hydrogel250E. coli and S. aureusRat modelFollowing 14 days of therapy, the test organism showed the slowest rate of re-epithelialization.[119]

7. Conclusions Challenges and Perspective

Wound healing with nanofibrous platforms loaded with silver nanoparticles has been studied biologically in vivo and in vitro as well as mechanically in this revisewarch. Because of their unique physicochemical and biological characteristics, AgNPs have drawn significant attention from researchers working on applications for wound healing. Ag nanoparticle-loaded electrospun nanofiber scaffolds also showed exceptional antibacterial activity, high porosity, non-toxicity, and biodegradability. Due to their hydrophilic qualities and prolonged release pattern, these nanofiber scaffolds have become more and more well-liked on a global scale. By promoting and hastening the healing process, they contribute significantly to wound dressing. Additionally, silver nanoparticles and other antibacterial substances together showed synergistic antibacterial properties. The effectiveness of silver nanoparticles in wound healing and skin regeneration has been established in numerous papers from various researchers. Without a doubt, additional research will produce scaffolds with unique properties beneficial for treating chronic wounds. However, since it avoids hazardous chemicals, manufacturing silver nanoparticles using green chemistry is an intelligent strategy. Silver nanoparticles can be produced through green chemistry and used for wound dressings to make them non-toxic and compatible with the body. Much research has been conducted on silver nanoparticles to improve the antibacterial capability of medical products such as wound dressings. Nonwoven mats made of electrospun nanofibers offer a great deal of potential for use in wound healing since they structurally resemble native extracellular matrix. Various biomedical applications, including leading-edge research aimed at healing chronic diabetic wounds, have investigated potential methods of nanotechnology-based medication delivery. In addition to their nanometric size, AgNPs were discovered to have possible use in treating diabetic wounds to lessen the likelihood of limb amputation. Their excellent antibacterial activity, anti-inflammatory response, and non-toxic nature make them an ideal and suitable alternative to other nanomaterials for wound dressing. AgNPs’ advantageous physicochemical characteristics support antibacterial effectiveness, and their surface charge also enables surface functionalization by coordinating particular ligands on the surface to try target-specific delivery. Consequential research has demonstrated the biocompatible AgNPs’ promise for treating diabetic wounds effectively, and a handful of the compounds have already been approved for commercialization. Several might be available soon with improved efficacy as an optimal dressing for successful wound healing in diabetes patients, according to a concurrent clinical study in human subjects. Additionally, those investigators found a combination of AgNPs and biopolymers to be more effective, and the inclusion of growth factors or phytochemicals may hasten wound healing by correcting any tissue damage. The exponential rise in research papers on green synthesis, when considering the AgNPs synthesis methods, is a drawback since it demonstrates the value and interest of plant materials in the production process. The synthesis rate of AgNPs is increased by using this economical and ecologically beneficial technique. However, morphological traits play a significant role in how effective AgNPs are. For the AgNPs to have the desired features, such as superior wound healing properties, it is necessary to standardize the optimization of the plant extracts and other reactive product characteristics. Additional research is required to link the physiological attributes of AgNPs with the physiological milieu in which they act. Before widespread usage in wound care products, careful consideration of their toxicity must be made because silver nanoparticles are incredibly active relative to their bulk. Extensive research into short- and long-term toxicity studies should be necessary to ascertain the underlying mechanism, and it should take thorough in vivo investigations into consideration as one of the future possibilities in developing AgNPs for wound healing. Additionally, special care must be given to the ideal AgNP dosage in formulations and suitable pairings to achieve a superior response in the diabetic wound. In recent years, nanotechnology has enabled the fabrication of various forms of AgNPs. However, AgNPs’ efficacy is hindered by their propensity for aggregation; surface passivator reagents are usually required to avoid accumulation. Further, silver oxidation may produce reactive oxygen species and radicals that can harm intracellular micro-organelles (such as mitochondria, ribosomes, and vacuoles) and macromolecules such as DNA, protein, and lipids. AgNPs are biocompatible but can occasionally result in argyria, according to research on how risk-free they are for people with DFU infections. However, most current studies on electrospun nanofibers throughout wound healing have been limited to pharmacodynamic assessments. As a result, the precise mechanism underlying nanofiber-assisted wound healing is unknown. Despite their increasing applications, comprehensive biological information still requires additional research due to several controversial results published on their safety. Researchers used chemical reduction methods to create a stable and colloidal dispersion of AgNPs using borohydride and hydrazine as reducing agents. Whereas these reduce the agent’s hyperactivity, they are toxic to the environment, limiting their applications.  

References

  1. Pugazhendhi, A.; Prabakar, D.; Jacob, J.M.; Karuppusamy, I.; Saratale, R.G. Synthesis and Characterization of Silver Nanoparticles Using Gelidium Amansii and Its Antimicrobial Property against Various Pathogenic Bacteria. Microb. Pathog. 2018, 114, 41–45.
  2. Lee, Y.H.; Cheng, F.Y.; Chiu, H.W.; Tsai, J.C.; Fang, C.Y.; Chen, C.W.; Wang, Y.J. Cytotoxicity, Oxidative Stress, Apoptosis and the Autophagic Effects of Silver Nanoparticles in Mouse Embryonic Fibroblasts. Biomaterials 2014, 35, 4706–4715.
  3. Cohen, M.S.; Stern, J.M.; Vanni, A.J.; Kelley, R.S.; Baumgart, E.; Field, D.; Libertino, J.A.; Summerhayes, I.C. In Vitro Analysis of a Nanocrystalline Silver-Coated Surgical Mesh. Surg. Infect. (Larchmt) 2007, 8, 397–403.
  4. Ibrahim, H.M.; Reda, M.M.; Klingner, A. Preparation and Characterization of Green Carboxymethylchitosan (CMCS)–Polyvinyl Alcohol (PVA) Electrospun Nanofibers Containing Gold Nanoparticles (AuNPs) and Its Potential Use as Biomaterials. Int. J. Biol. Macromol. 2020, 151, 821–829.
  5. Xu, Z.; Mahalingam, S.; Rohn, J.L.; Ren, G.; Edirisinghe, M. Physio-Chemical and Antibacterial Characteristics of Pressure Spun Nylon Nanofibres Embedded with Functional Silver Nanoparticles. Mater. Sci. Eng. C 2015, 56, 195–204.
  6. Munteanu, B.S.; Aytac, Z.; Pricope, G.M.; Uyar, T.; Vasile, C. Polylactic Acid (PLA)/Silver-NP/VitaminE Bionanocomposite Electrospun Nanofibers with Antibacterial and Antioxidant Activity. J. Nanoparticle Res. 2014, 16, 2643.
  7. Medici, S.; Peana, M.; Nurchi, V.M.; Zoroddu, M.A. Medical Uses of Silver: History, Myths, and Scientific Evidence. J. Med. Chem. 2019, 62, 5923–5943.
  8. Barillo, D.J.; Marx, D.E. Silver in Medicine: A Brief History BC 335 to Present. Burns 2014, 40, S3–S8.
  9. Franková, J.; Pivodová, V.; Vágnerová, H.; Juráňová, J.; Ulrichová, J. Effects of Silver Nanoparticles on Primary Cell Cultures of Fibroblasts and Keratinocytes in a Wound-Healing Model. J. Appl. Biomater. Funct. Mater. 2016, 14, e137–e142.
  10. You, C.; Li, Q.; Wang, X.; Wu, P.; Ho, J.K.; Jin, R.; Zhang, L.; Shao, H.; Han, C. Silver Nanoparticle Loaded Collagen/Chitosan Scaffolds Promote Wound Healing via Regulating Fibroblast Migration and Macrophage Activation. Sci. Rep. 2017, 7, 10489.
  11. Atiyeh, B.S.; Costagliola, M.; Hayek, S.N.; Dibo, S.A. Effect of Silver on Burn Wound Infection Control and Healing: Review of the Literature. Burns 2007, 33, 139–148.
  12. Rosli, N.A.; Teow, Y.H.; Mahmoudi, E. Current Approaches for the Exploration of Antimicrobial Activities of Nanoparticles. Sci. Technol. Adv. Mater. 2021, 22, 885–907.
  13. Gul, A.; Gallus, I.; Tegginamath, A.; Maryska, J.; Yalcinkaya, F. Electrospun Antibacterial Nanomaterials for Wound Dressings Applications. Membranes 2021, 11, 908.
  14. Wilkinson, L.J.; White, R.J.; Chipman, J.K. Silver and Nanoparticles of Silver in Wound Dressings: A Review of Efficacy and Safety. J. Wound Care 2011, 20, 543–549.
  15. Song, J.; Birbach, N.L.; Hinestroza, J.P. Deposition of Silver Nanoparticles on Cellulosic Fibers via Stabilization of Carboxymethyl Groups. Cellulose 2012, 19, 411–424.
  16. Lok, C.N.; Ho, C.M.; Chen, R.; He, Q.Y.; Yu, W.Y.; Sun, H.; Tam, P.K.H.; Chiu, J.F.; Che, C.M. Silver Nanoparticles: Partial Oxidation and Antibacterial Activities. J. Biol. Inorg. Chem. 2007, 12, 527–534.
  17. Kim, J.S.; Kuk, E.; Yu, K.N.; Kim, J.H.; Park, S.J.; Lee, H.J.; Kim, S.H.; Park, Y.K.; Park, Y.H.; Hwang, C.Y.; et al. Antimicrobial Effects of Silver Nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2007, 3, 95–101.
  18. Lara, H.H.; Garza-Treviño, E.N.; Ixtepan-Turrent, L.; Singh, D.K. Silver Nanoparticles Are Broad-Spectrum Bactericidal and Virucidal Compounds. J. Nanobiotechnol. 2011, 9, 30.
  19. Galdiero, S.; Falanga, A.; Vitiello, M.; Cantisani, M.; Marra, V.; Galdiero, M. Silver Nanoparticles as Potential Antiviral Agents. Molecules 2011, 16, 8894–8918.
  20. Zorraquín-Peña, I.; Cueva, C.; de Llano, D.G.; Bartolomé, B.; Moreno-Arribas, M.V. Glutathione-Stabilized Silver Nanoparticles: Antibacterial Activity against Periodontal Bacteria, and Cytotoxicity and Inflammatory Response in Oral Cells. Biomedicines 2020, 8, 375.
  21. Pal, S.; Nisi, R.; Stoppa, M.; Licciulli, A. Silver-Functionalized Bacterial Cellulose as Antibacterial Membrane for Wound-Healing Applications. ACS Omega 2017, 2, 3632–3639.
  22. Konop, M.; Damps, T.; Misicka, A.; Rudnicka, L. Certain Aspects of Silver and Silver Nanoparticles in Wound Care: A Minireview. J. Nanomater. 2016, 2016, 7614753.
  23. You, C.; Han, C.; Wang, X.; Zheng, Y.; Li, Q.; Hu, X.; Sun, H. The Progress of Silver Nanoparticles in the Antibacterial Mechanism, Clinical Application and Cytotoxicity. Mol. Biol. Rep. 2012, 39, 9193–9201.
  24. Shanmuganathan, R.; Karuppusamy, I.; Saravanan, M.; Muthukumar, H.; Ponnuchamy, K.; Ramkumar, V.S.; Pugazhendhi, A. Synthesis of Silver Nanoparticles and Their Biomedical Applications-A Comprehensive Review. Curr. Pharm. Des. 2019, 25, 2650–2660.
  25. Srikar, S.K.; Giri, D.D.; Pal, D.B.; Mishra, P.K.; Upadhyay, S.N. Green Synthesis of Silver Nanoparticles: A Review. Green Sustain. Chem. 2016, 6, 34–56.
  26. Rajendran, N.K.; Kumar, S.S.D.; Houreld, N.N.; Abrahamse, H. A Review on Nanoparticle Based Treatment for Wound Healing. J. Drug Deliv. Sci. Technol. 2018, 44, 421–430.
  27. Berthet, M.; Gauthier, Y.; Lacroix, C.; Verrier, B.; Monge, C. Nanoparticle-Based Dressing: The Future of Wound Treatment? Trends Biotechnol. 2017, 35, 770–784.
  28. Wang, Y.; Qiao, Y.; Wang, P.; Li, Q.; Xia, C.; Ju, M. Bio Fabrication of Silver Nanoparticles as an Effective Wound Healing Agent in the Wound Care after Anorectal Surgery. J. Photochem. Photobiol. B Biol. 2018, 178, 457–462.
  29. Ahlberg, S.; Meinke, M.C.; Werner, L.; Epple, M.; Diendorf, J.; Blume-Peytavi, U.; Lademann, J.; Vogt, A.; Rancan, F. Comparison of Silver Nanoparticles Stored under Air or Argon with Respect to the Induction of Intracellular Free Radicals and Toxic Effects toward Keratinocytes. Eur. J. Pharm. Biopharm. 2014, 88, 651–657.
  30. Zhang, X.F.; Liu, Z.G.; Shen, W.; Gurunathan, S. Silver Nanoparticles: Synthesis, Characterization, Properties, Applications, and Therapeutic Approaches. Int. J. Mol. Sci. 2016, 17, 1534.
  31. Hebeish, A.; El-Rafie, M.H.; EL-Sheikh, M.A.; Seleem, A.A.; El-Naggar, M.E. Antimicrobial Wound Dressing and Anti-Inflammatory Efficacy of Silver Nanoparticles. Int. J. Biol. Macromol. 2014, 65, 509–515.
  32. Kim, K.J.; Sung, W.S.; Suh, B.K.; Moon, S.K.; Choi, J.S.; Kim, J.G.; Lee, D.G. Antifungal Activity and Mode of Action of Silver Nano-Particles on Candida Albicans. BioMetals 2009, 22, 235–242.
  33. Gajbhiye, M.; Kesharwani, J.; Ingle, A.; Gade, A.; Rai, M. Fungus-Mediated Synthesis of Silver Nanoparticles and Their Activity against Pathogenic Fungi in Combination with Fluconazole. Nanomed. Nanotechnol. Biol. Med. 2009, 5, 382–386.
  34. Jo, Y.K.; Kim, B.H.; Jung, G. Antifungal Activity of Silver Ions and Nanoparticles on Phytopathogenic Fungi. Plant Dis. 2009, 93, 1037–1043.
  35. Speshock, J.L.; Murdock, R.C.; Braydich-Stolle, L.K.; Schrand, A.M.; Hussain, S.M. Interaction of Silver Nanoparticles with Tacaribe Virus. J. Nanobiotechnol. 2010, 8, 19.
  36. Narasimha, G. Antiviral Activity of Silver Nanoparticles Synthesized by Fungal Strain Aspergillus Niger. J. Nanosci. Nanotechnol. 2012, 6, 18–20.
  37. Alshabanah, L.A.; Hagar, M.; Al-Mutabagani, L.A.; Abozaid, G.M.; Abdallah, S.M.; Shehata, N.; Ahmed, H.; Hassanin, A.H. Hybrid Nanofibrous Membranes as a Promising Functional Layer for Personal Protection Equipment: Manufacturing and Antiviral/Antibacterial Assessments. Polymers 2021, 13, 1776.
  38. Lokina, S.; Stephen, A.; Kaviyarasan, V.; Arulvasu, C.; Narayanan, V. Cytotoxicity and Antimicrobial Activities of Green Synthesized Silver Nanoparticles. Eur. J. Med. Chem. 2014, 76, 256–263.
  39. Sankar, R.; Karthik, A.; Prabu, A.; Karthik, S.; Shivashangari, K.S.; Ravikumar, V. Origanum Vulgare Mediated Biosynthesis of Silver Nanoparticles for Its Antibacterial and Anticancer Activity. Colloids Surf. B Biointerfaces 2013, 108, 80–84.
  40. Vasanth, K.; Ilango, K.; MohanKumar, R.; Agrawal, A.; Dubey, G.P. Anticancer Activity of Moringa Oleifera Mediated Silver Nanoparticles on Human Cervical Carcinoma Cells by Apoptosis Induction. Colloids Surf. B Biointerfaces 2014, 117, 354–359.
  41. Guo, D.; Dou, D.; Ge, L.; Huang, Z.; Wang, L.; Gu, N. A Caffeic Acid Mediated Facile Synthesis of Silver Nanoparticles with Powerful Anti-Cancer Activity. Colloids Surf.B Biointerfaces 2015, 134, 229–234.
  42. Venkatesan, J.; Lee, J.Y.; Kang, D.S.; Anil, S.; Kim, S.K.; Shim, M.S.; Kim, D.G. Antimicrobial and Anticancer Activities of Porous Chitosan-Alginate Biosynthesized Silver Nanoparticles. Int. J. Biol. Macromol. 2017, 98, 515–525.
  43. Salahuddin, N.; Elbarbary, A.A.; Alkabes, H.A. Antibacterial and Anticancer Activity of Loaded Quinazolinone Polypyrrole/Chitosan Silver Chloride Nanocomposite. Int. J. Polym. Mater. Polym. Biomater. 2017, 66, 307–316.
  44. El-Sherbiny, I.M.; Salih, E.; Yassin, A.M.; Hafez, E.E. Newly Developed Chitosan-Silver Hybrid Nanoparticles: Biosafety and Apoptosis Induction in HepG2 Cells. J. Nanoparticle Res. 2016, 18, 172.
  45. Lu, W.; Senapati, D.; Wang, S.; Tovmachenko, O.; Singh, A.K.; Yu, H.; Ray, P.C. Effect of Surface Coating on the Toxicity of Silver Nanomaterials on Human Skin Keratinocytes. Chem. Phys. Lett. 2010, 487, 92–96.
  46. Palanki, R.; Arora, S.; Tyagi, N.; Rusu, L.; Singh, A.P.; Palanki, S.; Carter, J.E.; Singh, S. Size Is an Essential Parameter in Governing the UVB-Protective Efficacy of Silver Nanoparticles in Human Keratinocytes. BMC Cancer 2015, 15, 636.
  47. Kamareddine, L. The Biological Control of the Malaria Vector. Toxins 2012, 4, 748–767.
  48. Jiang, Z.; Jiang, D.; Showkot Hossain, A.M.; Qian, K.; Xie, J. In Situ Synthesis of Silver Supported Nanoporous Iron Oxide Microbox Hybrids from Metal-Organic Frameworks and Their Catalytic Application in p-Nitrophenol Reduction. Phys. Chem. Chem. Phys. 2015, 17, 2550–2559.
  49. Ajitha, B.; Ashok Kumar Reddy, Y.; Shameer, S.; Rajesh, K.M.; Suneetha, Y.; Sreedhara Reddy, P. Lantana Camara Leaf Extract Mediated Silver Nanoparticles: Antibacterial, Green Catalyst. J. Photochem. Photobiol. B Biol. 2015, 149, 84–92.
  50. Bergin, S.; Wraight, P. Silver Based Wound Dressings and Topical Agents for Treating Diabetic Foot Ulcers. Cochrane Database Syst. Rev. 2006, CD005082.
  51. Liu, X.; Lee, P.Y.; Ho, C.M.; Lui, V.C.H.; Chen, Y.; Che, C.M.; Tam, P.K.H.; Wong, K.K.Y. Silver Nanoparticles Mediate Differential Responses in Keratinocytes and Fibroblasts during Skin Wound Healing. ChemMedChem 2010, 5, 468–475.
  52. Nam, G.; Rangasamy, S.; Purushothaman, B.; Song, J.M. The Application of Bactericidal Silver Nanoparticles in Wound Treatment. Nanomater. Nanotechnol. 2015, 5, 5–23.
  53. Ai , J.; Biazar, E.; Jafarpour, M.; Montazeri, M.; Majdi, A.; Aminifard, S.; Zafari, M.; Akbari, H.R.; Rad, H.G. Nanotoxicology and Nanoparticle Safety in Biomedical Designs. Int. J. Nanomed. 2011, 6, 1117.
  54. Chakrabarti, S.; Chattopadhyay, P.; Islam, J.; Ray, S.; Raju, P.S.; Mazumder, B. Aspects of Nanomaterials in Wound Healing. Curr. Drug Deliv. 2018, 16, 26–41.
  55. Tyavambiza, C.; Elbagory, A.M.; Madiehe, A.M.; Meyer, M.; Meyer, S. The Antimicrobial and Anti-Inflammatory Effects of Silver Nanoparticles Synthesised from Cotyledon Orbiculata Aqueous Extract. Nanomaterials 2021, 11, 1343.
  56. Jura, J.; Szmyd, R.; Goralczyk, A.G.; Skalniak, L.; Cierniak, A.; Lipert, B.; Filon, F.L.; Crosera, M.; Borowczyk, J.; Laczna, E.; et al. Effect of Silver Nanoparticles on Human Primary Keratinocytes. Biol. Chem. 2013, 394, 113–123.
  57. Gallo, A.L.; Paladini, F.; Romano, A.; Verri, T.; Quattrini, A.; Sannino, A.; Pollini, M. Efficacy of Silver Coated Surgical Sutures on Bacterial Contamination, Cellular Response and Wound Healing. Mater. Sci. Eng. C 2016, 69, 884–893.
  58. Paladini, F.; Picca, R.A.; Sportelli, M.C.; Cioffi, N.; Sannino, A.; Pollini, M. Surface Chemical and Biological Characterization of Flax Fabrics Modified with Silver Nanoparticles for Biomedical Applications. Mater. Sci. Eng. C 2015, 52, 1–10.
  59. Paladini, F.; De Simone, S.; Sannino, A.; Pollini, M. Antibacterial and Antifungal Dressings Obtained by Photochemical Deposition of Silver Nanoparticles. J. Appl. Polym. Sci. 2014, 131, 40326.
  60. Tian, J.; Wong, K.K.Y.; Ho, C.M.; Lok, C.N.; Yu, W.Y.; Che, C.M.; Chiu, J.F.; Tam, P.K.H. Topical Delivery of Silver Nanoparticles Promotes Wound Healing. ChemMedChem 2007, 2, 129–136.
  61. Meier, B.; Radeke, H.H.; Selle, S.; Younes, M.; Sies, H.; Resch, K.; Habermehl, G.G. Human Fibroblasts Release Reactive Oxygen Species in Response to Interleukin-1 or Tumour Necrosis Factor-α. Biochem. J. 1989, 263, 539–545.
  62. Yu, K.; Zhou, X.; Zhu, T.; Wu, T.; Wang, J.; Fang, J.; El-Aassar, M.R.; El-Hamshary, H.; El-Newehy, M.; Mo, X. Fabrication of Poly(Ester-Urethane)Urea Elastomer/Gelatin Electrospun Nanofibrous Membranes for Potential Applications in Skin Tissue Engineering. RSC Adv. 2016, 6, 73636–73644.
  63. Burdușel, A.C.; Gherasim, O.; Grumezescu, A.M.; Mogoantă, L.; Ficai, A.; Andronescu, E. Biomedical Applications of Silver Nanoparticles: An up-to-Date Overview. Nanomaterials 2018, 8, 681.
  64. Baygar, T.; Sarac, N.; Ugur, A.; Karaca, I.R. Antimicrobial Characteristics and Biocompatibility of the Surgical Sutures Coated with Biosynthesized Silver Nanoparticles. Bioorg. Chem. 2019, 86, 254–258.
  65. Prabhu, S.; Poulose, E.K. Silver Nanoparticles: Mechanism of Antimicrobial Action, Synthesis, Medical Applications, and Toxicity Effects. Int. Nano Lett. 2012, 2, 32.
  66. Calamak, S.; Aksoy, E.A.; Ertas, N.; Erdogdu, C.; Sagıroglu, M.; Ulubayram, K. Ag/Silk Fibroin Nanofibers: Effect of Fibroin Morphology on Ag+ Release and Antibacterial Activity. Eur. Polym. J. 2015, 67, 99–112.
  67. Gaillet, S.; Rouanet, J.M. Silver Nanoparticles: Their Potential Toxic Effects after Oral Exposure and Underlying Mechanisms-A Review. Food Chem. Toxicol. 2015, 77, 58–63.
  68. Lansdown, A.B.G. A Pharmacological and Toxicological Profile of Silver as an Antimicrobial Agent in Medical Devices. Adv. Pharmacol. Sci. 2010, 2010, 910686.
  69. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano Based Drug Delivery Systems: Recent Developments and Future Prospects 10 Technology 1007 Nanotechnology 03 Chemical Sciences 0306 Physical Chemistry (Incl. Structural) 03 Chemical Sciences 0303 Macromolecular and Materials Chemistry 11 Medical and He. J. Nanobiotechnol. 2018, 16, 71.
  70. Yin, I.X.; Zhang, J.; Zhao, I.S.; Mei, M.L.; Li, Q.; Chu, C.H. The Antibacterial Mechanism of Silver Nanoparticles and Its Application in Dentistry. Int. J. Nanomed. 2020, 15, 2555–2562.
  71. Dai, X.; Guo, Q.; Zhao, Y.; Zhang, P.; Zhang, T.; Zhang, X.; Li, C. Functional Silver Nanoparticle as a Benign Antimicrobial Agent That Eradicates Antibiotic-Resistant Bacteria and Promotes Wound Healing. ACS Appl. Mater. Interfaces 2016, 8, 25798–25807.
  72. Deng, H.; McShan, D.; Zhang, Y.; Sinha, S.S.; Arslan, Z.; Ray, P.C.; Yu, H. Mechanistic Study of the Synergistic Antibacterial Activity of Combined Silver Nanoparticles and Common Antibiotics. Environ. Sci. Technol. 2016, 50, 8840–8848.
  73. Roy, A.; Bulut, O.; Some, S.; Mandal, A.K.; Yilmaz, M.D. Green Synthesis of Silver Nanoparticles: Biomolecule-Nanoparticle Organizations Targeting Antimicrobial Activity. RSC Adv. 2019, 9, 2673–2702.
  74. Salleh, A.; Naomi, R.; Utami, N.D.; Mohammad, A.W.; Mahmoudi, E.; Mustafa, N.; Fauzi, M.B. The Potential of Silver Nanoparticles for Antiviral and Antibacterial Applications: A Mechanism of Action. Nanomaterials 2020, 10, 1556.
  75. Hasan, S. A Review on Nanoparticles : Their Synthesis and Types. Res. J. Recent Sci. Res. J. Recent. Sci. Uttar Pradesh (Lucknow Campus) 2014, 4, 1–3.
  76. Khan, A.U.; Malik, N.; Khan, M.; Cho, M.H.; Khan, M.M. Fungi-Assisted Silver Nanoparticle Synthesis and Their Applications. Bioprocess Biosyst. Eng. 2018, 41, 1–20.
  77. Sintubin, L.; Verstraete, W.; Boon, N. Biologically Produced Nanosilver: Current State and Future Perspectives. Biotechnol. Bioeng. 2012, 109, 2422–2436.
  78. Singh, P.; Kim, Y.J.; Zhang, D.; Yang, D.C. Biological Synthesis of Nanoparticles from Plants and Microorganisms. Trends Biotechnol. 2016, 34, 588–599.
  79. Aboyewa, J.A.; Sibuyi, N.R.S.; Meyer, M.; Oguntibeju, O.O. Green Synthesis of Metallic Nanoparticles Using Some Selected Medicinal Plants from Southern Africa and Their Biological Applications. Plants 2021, 10, 1929.
  80. Simon, S.; Sibuyi, N.R.S.; Fadaka, A.O.; Meyer, M.; Madiehe, A.M.; du Preez, M.G. The Antimicrobial Activity of Biogenic Silver Nanoparticles Synthesized from Extracts of Red and Green European Pear Cultivars. Artif. Cells 2021, 49, 614–625.
  81. Rajan, R.; Chandran, K.; Harper, S.L.; Yun, S.-I.; Kalaichelvan, P.T. Plant Extract Synthesized Silver Nanoparticles: An Ongoing Source of Novel Biocompatible Materials. Ind. Crops Prod. 2015, 70, 356–373.
  82. Ahmed, S.; Ahmad, M.; Swami, B.L.; Ikram, S. A Review on Plants Extract Mediated Synthesis of Silver Nanoparticles for Antimicrobial Applications: A Green Expertise. J. Adv. Res. 2016, 7, 17–28.
  83. Kumar, S.; Basumatary, I.B.; Sudhani, H.P.K.; Bajpai, V.K.; Chen, L.; Shukla, S.; Mukherjee, A. Plant Extract Mediated Silver Nanoparticles and Their Applications as Antimicrobials and in Sustainable Food Packaging: A State-of-the-Art Review. Trends Food Sci. Technol. 2021, 112, 651–666.
  84. Patel, P.; Agarwal, P.; Kanawaria, S.; Kachhwaha, S.; Kothari, S.L. Plant-Based Synthesis of Silver Nanoparticles and Their Characterization. Nanotechnol. Plant Sci. Nanoparticles Their Impact Plants 2015, 271–288.
  85. Agarwal, Y.; Rajinikanth, P.S.; Ranjan, S.; Tiwari, U.; Balasubramnaiam, J.; Pandey, P.; Arya, D.K.; Anand, S.; Deepak, P. Curcumin Loaded Polycaprolactone-/Polyvinyl Alcohol-Silk Fibroin Based Electrospun Nanofibrous Mat for Rapid Healing of Diabetic Wound: An in-Vitro and in-Vivo Studies. Int. J. Biol. Macromol. 2021, 176, 376–386.
  86. Greiner, A.; Wendorff, J.H. Electrospinning: A Fascinating Method for the Preparation of Ultrathin Fibers. Angew. Chemie-Int. Ed. 2007, 46, 5670–5703.
  87. Kadavil, H.; Zagho, M.; Elzatahry, A.; Altahtamouni, T. Sputtering of Electrospun Polymer-Based Nanofibers for Biomedical Applications: A Perspective. Nanomaterials 2019, 9, 77.
  88. Zhang, Y.; Li, S.; Xu, Y.; Shi, X.; Zhang, M.; Huang, Y.; Liang, Y.; Chen, Y.; Ji, W.; Kim, J.R.; et al. Engineering of Hollow Polymeric Nanosphere-Supported Imidazolium-Based Ionic Liquids with Enhanced Antimicrobial Activities. Nano Res. 2022, 15, 5556–5568.
  89. Tang, Y.; Varyambath, A.; Ding, Y.; Chen, B.; Huang, X.; Zhang, Y.; Yu, D.G.; Kim, I.; Song, W. Porous Organic Polymers for Drug Delivery: Hierarchical Pore Structures, Variable Morphologies, and Biological Properties. Biomater. Sci. 2022.
  90. Zhang, Y.; Kim, I.; Lu, Y.; Xu, Y.; Yu, D.G.; Song, W. Intelligent Poly(L-Histidine)-Based Nanovehicles for Controlled Drug Delivery. J. Control. Release 2022, 349, 963–982.
  91. Kenry; Lim, C.T. Nanofiber Technology: Current Status and Emerging Developments. Prog. Polym. Sci. 2017, 70, 1–17.
  92. Morgado, P.I.; Aguiar-Ricardo, A.; Correia, I.J. Asymmetric Membranes as Ideal Wound Dressings: An Overview on Production Methods, Structure, Properties and Performance Relationship. J. Memb. Sci. 2015, 490, 139–151.
  93. Souto, E.B.; Yoshida, C.M.P.; Leonardi, G.R.; Cano, A.; Sanchez-Lopez, E.; Zielinska, A.; Viseras, C.; Severino, P.; da Silva, C.F.; Barbosa, R.d.M. Lipid-Polymeric Films: Composition, Production and Applications in Wound Healing and Skin Repair. Pharmaceutics 2021, 13, 1199.
  94. Dumville, J.C.; Deshpande, S.; O’Meara, S.; Speak, K. Hydrocolloid Dressings for Healing Diabetic Foot Ulcers. Cochrane Database Syst. Rev. 2013, 2013, CD009099.
  95. Liang, Y.; He, J.; Guo, B. Functional Hydrogels as Wound Dressing to Enhance Wound Healing. ACS Nano 2021, 15, 12687–12722.
  96. Henry, L.A.; Hart, M. Regeneration from Injury and Resource Allocation in Sponges and Corals-A Review. Int. Rev. Hydrobiol. 2005, 90, 125–158.
  97. Archana, D.; Singh, B.K.; Dutta, J.; Dutta, P.K. Chitosan-PVP-Nano Silver Oxide Wound Dressing: In Vitro and in Vivo Evaluation. Int. J. Biol. Macromol. 2015, 73, 49–57.
  98. Duraipandy, N.; Lakra, R.; Vinjimur Srivatsan, K.; Ramamoorthy, U.; Korrapati, P.S.; Kiran, M.S. Plumbagin Caged Silver Nanoparticle Stabilized Collagen Scaffold for Wound Dressing. J. Mater. Chem. B 2015, 3, 1415–1425.
  99. An, J.; Zhang, H.; Zhang, J.; Zhao, Y.; Yuan, X. Preparation and Antibacterial Activity of Electrospun Chitosan/ Poly(Ethylene Oxide) Membranes Containing Silver Nanoparticles. Colloid Polym. Sci. 2009, 287, 1425–1434.
  100. Madhumathi, K.; Sudheesh Kumar, P.T.; Abhilash, S.; Sreeja, V.; Tamura, H.; Manzoor, K.; Nair, S.V.; Jayakumar, R. Development of Novel Chitin/Nanosilver Composite Scaffolds for Wound Dressing Applications. J. Mater. Sci. Mater. Med. 2010, 21, 807–813.
  101. Kim, M.H.; Cho, D.; Kwon, O.H.; Park, W.H. Thermal Fabrication and Characterization of Ag Nanoparticle-Activated Carbon Composites for Functional Wound-Dressing Additives. J. Alloys Compd. 2018, 735, 2670–2674.
  102. Paladini, F.; Di Franco, C.; Panico, A.; Scamarcio, G.; Sannino, A.; Pollini, M. In Vitro Assessment of the Antibacterial Potential of Silver Nano-Coatings on Cotton Gauzes for Prevention of Wound Infections. Materials 2016, 9, 411.
  103. Namviriyachote, N.; Lipipun, V.; Akkhawattanangkul, Y.; Charoonrut, P.; Ritthidej, G.C. Development of Polyurethane Foam Dressing Containing Silver and Asiaticoside for Healing of Dermal Wound. Asian J. Pharm. Sci. 2019, 14, 63–77.
  104. Rattanaruengsrikul, V.; Pimpha, N.; Supaphol, P. In Vitro Efficacy and Toxicology Evaluation of Silver Nanoparticle-Loaded Gelatin Hydrogel Pads as Antibacterial Wound Dressings. J. Appl. Polym. Sci. 2012, 124, 1668–1682.
  105. Masood, N.; Ahmed, R.; Tariq, M.; Ahmed, Z.; Masoud, M.S.; Ali, I.; Asghar, R.; Andleeb, A.; Hasan, A. Silver Nanoparticle Impregnated Chitosan-PEG Hydrogel Enhances Wound Healing in Diabetes Induced Rabbits. Int. J. Pharm. 2019, 559, 23–36.
  106. Chen, M.; Yang, Z.; Wu, H.; Pan, X.; Xie, X.; Wu, C. Antimicrobial Activity and the Mechanism of Silver Nanoparticle Thermosensitive Gel. Int. J. Nanomed. 2011, 6, 2873–2877.
  107. Shao, J.; Wang, B.; Li, J.; Jansen, J.A.; Walboomers, X.F.; Yang, F. Antibacterial Effect and Wound Healing Ability of Silver Nanoparticles Incorporation into Chitosan-Based Nanofibrous Membranes. Mater. Sci. Eng. C 2019, 98, 1053–1063.
  108. Liang, D.; Lu, Z.; Yang, H.; Gao, J.; Chen, R. Novel Asymmetric Wettable AgNPs/Chitosan Wound Dressing: In Vitro and in Vivo Evaluation. ACS Appl. Mater. Interfaces 2016, 8, 3958–3968.
  109. Ye, D.; Zhong, Z.; Xu, H.; Chang, C.; Yang, Z.; Wang, Y.; Ye, Q.; Zhang, L. Construction of Cellulose/Nanosilver Sponge Materials and Their Antibacterial Activities for Infected Wounds Healing. Cellulose 2016, 23, 749–763.
  110. Pérez-Díaz, M.; Alvarado-Gomez, E.; Magaña-Aquino, M.; Sánchez-Sánchez, R.; Velasquillo, C.; Gonzalez, C.; Ganem-Rondero, A.; Martínez-Castañon, G.; Zavala-Alonso, N.; Martinez-Gutierrez, F. Anti-Biofilm Activity of Chitosan Gels Formulated with Silver Nanoparticles and Their Cytotoxic Effect on Human Fibroblasts. Mater. Sci. Eng. C 2016, 60, 317–323.
  111. Pei, Z.; Sun, Q.; Sun, X.; Wang, Y.; Zhao, P. Preparation and Characterization of Silver Nanoparticles on Silk Fibroin/Carboxymethy Lchitosan Composite Sponge as Anti-Bacterial Wound Dressing. Biomed. Mater. Eng. 2015, 26, S111–S118.
  112. Ding, L.; Shan, X.; Zhao, X.; Zha, H.; Chen, X.; Wang, J.; Cai, C.; Wang, X.; Li, G.; Hao, J.; et al. Spongy Bilayer Dressing Composed of Chitosan–Ag Nanoparticles and Chitosan–Bletilla Striata Polysaccharide for Wound Healing Applications. Carbohydr. Polym. 2017, 157, 1538–1547.
  113. Wu, J.; Zheng, Y.; Song, W.; Luan, J.; Wen, X.; Wu, Z.; Chen, X.; Wang, Q.; Guo, S. In Situ Synthesis of Silver-Nanoparticles/Bacterial Cellulose Composites for Slow-Released Antimicrobial Wound Dressing. Carbohydr. Polym. 2014, 102, 762–771.
  114. Wu, J.; Zheng, Y.; Wen, X.; Lin, Q.; Chen, X.; Wu, Z. Silver Nanoparticle/Bacterial Cellulose Gel Membranes for Antibacterial Wound Dressing: Investigation in Vitro and in Vivo. Biomed. Mater. 2014, 9, 035005.
  115. Kumar, P.T.S.; Abhilash, S.; Manzoor, K.; Nair, S.V.; Tamura, H.; Jayakumar, R. Preparation and Characterization of Novel β-Chitin/Nanosilver Composite Scaffolds for Wound Dressing Applications. Carbohydr. Polym. 2010, 80, 761–767.
  116. Montaser, A.S.; Abdel-Mohsen, A.M.; Ramadan, M.A.; Sleem, A.A.; Sahffie, N.M.; Jancar, J.; Hebeish, A. Preparation and Characterization of Alginate/Silver/Nicotinamide Nanocomposites for Treating Diabetic Wounds. Int. J. Biol. Macromol. 2016, 92, 739–747.
  117. Abdel-Mohsen, A.M.; Hrdina, R.; Burgert, L.; Abdel-Rahman, R.M.; Hašová, M.; Šmejkalová, D.; Kolář, M.; Pekar, M.; Aly, A.S. Antibacterial Activity and Cell Viability of Hyaluronan Fiber with Silver Nanoparticles. Carbohydr. Polym. 2013, 92, 1177–1187.
  118. Lu, Z.; Gao, J.; He, Q.; Wu, J.; Liang, D.; Yang, H.; Chen, R. Enhanced Antibacterial and Wound Healing Activities of Microporous Chitosan-Ag/ZnO Composite Dressing. Carbohydr. Polym. 2017, 156, 460–469.
  119. Verma, J.; Kanoujia, J.; Parashar, P.; Tripathi, C.B.; Saraf, S.A. Wound Healing Applications of Sericin/Chitosan-Capped Silver Nanoparticles Incorporated Hydrogel. Drug Deliv. Transl. Res. 2017, 7, 77–88.
More